1
|
Chien HW, Chuang CC, Hsieh YH, Lee CY, Yu NY, Yang SF. Tricetin suppresses the cell migration and BMP-6 expression through p38 signaling pathways in human retinal pigment epithelium cells. ENVIRONMENTAL TOXICOLOGY 2024; 39:4148-4155. [PMID: 38654487 DOI: 10.1002/tox.24294] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Subscribe] [Scholar Register] [Received: 01/10/2024] [Revised: 01/29/2024] [Accepted: 02/10/2024] [Indexed: 04/26/2024]
Abstract
Proliferative vitreoretinopathy (PVR) is a visual-threatening disease, which cause from the migration of retinal pigment epithelium (RPE). Tricetin, a family of flavonoids, can inhibit the metastasis of several cancers. Herein, we aim to evaluate the possible effect of tricetin on inhibiting ARPE-19 cells migration. The Boyden chamber assay, wound healing assay, RNA sequencing, and Western blot analysis were applied in our experiment. The results revealed that tricetin inhibited the cell migration abilities of ARPE-19 cells. Moreover, using RNA sequencing technology, we revealed that tricetin repressed bone morphogenetic protein-6 (BMP-6) gene expressions in ARPE-19 cells. Overexpression of BMP-6 resulted in significant restoration of cell migration capabilities of tricetin-treated ARPE-19 cells. Furthermore, tricetin suppressed the phosphorylation of the p38 signaling pathway. Moreover, blocking the p38 pathway also inhibits BMP-6 expression and migration in the ARPE-19 cells. In conclusion, this study revealed that tricetin inhibits the ARPE-19 cell migration mainly via the suppression of BMP-6 expression and p38 signaling pathway.
Collapse
Affiliation(s)
- Hsiang-Wen Chien
- Department of Ophthalmology, Cathay General Hospital, Taipei, Taiwan
- Department of Ophthalmology, Sijhih Cathay General Hospital, New Taipei City, Taiwan
- School of Medicine, National Tsing Hua University, Hsinchu, Taiwan
- School of Medicine, College of Medicine, Fu Jen Catholic University, New Taipei, Taiwan
| | - Chih-Chun Chuang
- Department of Ophthalmology, Changhua Christian Hospital, Changhua, Taiwan
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Post-Baccalaureate Medicine, College of Medicine, National Chung Hsing University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chia-Yi Lee
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Nobel Eye Institute, Taipei, Taiwan
| | - Nuo-Yi Yu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
2
|
Sui L, Cong Y, Liu M, Liu X, Xu Y, Jiang WG, Ye L. Upregulated bone morphogenetic protein 8A (BMP8A) in triple negative breast cancer (TNBC) and its involvement in the bone metastasis. Front Cell Dev Biol 2024; 12:1374269. [PMID: 39100096 PMCID: PMC11294076 DOI: 10.3389/fcell.2024.1374269] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 01/21/2024] [Accepted: 06/26/2024] [Indexed: 08/06/2024] Open
Abstract
Objective The present study aimed to investigate the involvement of aberrant BMP8A expression in TNBC and bone metastasis. Methods Aberrant expression of BMP8A in breast cancer was first determined by analyzing The Cancer Genome Atlas breast cancer cohort (TCGA-BRCA) and an immunohistochemical (IHC) staining of BMP8A in a breast cancer tissue microarray (TMA). Clinical relevance of deregulated BMP8A in breast cancer was assessed using Kaplan-Meier online analysis. The influence of BMP8A on cellular functions of two TNBC cell lines was assessed using in vitro assays. Conditional medium (CM) collected from the supernatant of hFOB cells and bone matrix extract (BME) was applied to mimic the bone micro-environment to evaluate the role played by BMP8A in bone metastasis. Correlations with both osteolytic and osteoblastic markers were evaluated in the TCGA-BRCA cohort. Expression of certain responsive genes was quantified in the BMP8A overexpression cell lines. Additionally, signal transduction through both Smad-dependent and independent pathways was evaluated using Western blot assay. Results Compared to the adjacent normal tissues, BMP8A expression was significantly increased in primary tumors (p < 0.05) which was associated with shorter distant metastasis free survival (DMFS) in TNBC (p < 0.05). BMP8A was observed to enhance cell invasion and migration within TNBC cells. In the simulated bone milieu, both MDA-MB-231BMP8Aexp and BT549BMP8Aexp cells presented enhanced invasiveness. BMP8A level was strongly correlated with most osteolytic and osteoblastic markers, suggesting the potential involvement of BMP8A in bone metastasis in TNBC. Receptor activator of nuclear factor kappa-B ligand (RANKL) expression was significantly increased in BMP8A overexpressed triple-negative cell lines (MDA-MB-231 and BT549). Furthermore, enhanced phosphorylation of Smad3 and increased expression of epidermal growth factor receptor (EGFR) were observed in MDA-MB-231 cells overexpressing BMP8A. Conclusion BMP8A was upregulated in TNBC which was associated with poorer DMFS. BMP8A overexpression enhanced the invasion and migration of TNBC cells. With a putative role in osteolytic bone metastasis in TNBC, BMP8A represents a promising candidate for further investigation into its therapeutic potential.
Collapse
Affiliation(s)
- Laijian Sui
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
- Department of Orthopedics, Yantai Yuhuangding Hospital, Yantai, Shandong, China
| | - Yizi Cong
- Department of Breast Surgery, Yantai Yuhuangding Hospital, Qingdao University, Yantai, China
| | - Ming Liu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Xiangyi Liu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Yali Xu
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Wen G. Jiang
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| | - Lin Ye
- Cardiff China Medical Research Collaborative, Division of Cancer and Genetics, Cardiff University School of Medicine, Cardiff, United Kingdom
| |
Collapse
|
3
|
Tanabe H, Suzuki T, Ohishi T, Isemura M, Nakamura Y, Unno K. Effects of Epigallocatechin-3-Gallate on Matrix Metalloproteinases in Terms of Its Anticancer Activity. MOLECULES (BASEL, SWITZERLAND) 2023; 28:molecules28020525. [PMID: 36677584 PMCID: PMC9862901 DOI: 10.3390/molecules28020525] [Citation(s) in RCA: 15] [Impact Index Per Article: 7.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Figures] [Subscribe] [Scholar Register] [Received: 11/22/2022] [Revised: 12/29/2022] [Accepted: 12/31/2022] [Indexed: 01/06/2023]
Abstract
Epidemiological studies have shown that the consumption of green tea has beneficial effects against cancer. Basic studies have provided evidence that epigallocatechin gallate (EGCG) is a major contributor to these effects. Matrix metalloproteinases (MMPs) are zinc-dependent metalloproteinases with the ability to degrade the extracellular matrix proteins and are involved in various diseases including cancer in which MMPs have a critical role in invasion and metastasis. In this review, we discuss the effects of EGCG on several types of MMPs in the context of its anticancer activity. In the promoter region, MMPs have binding sites for at least one transcription factor of AP-1, Sp1, and NF-κB, and EGCG can downregulate these transcription factors through signaling pathways mediated by reactive oxygen species. EGCG can also decrease nuclear ERK, p38, heat shock protein-27 (Hsp27), and β-catenin levels, leading to suppression of MMPs' expression. Other mechanisms by which EGCG inhibits MMPs include direct binding to MMPs to prevent their activation and downregulation of NF-κB to suppress the production of inflammatory cytokines such as TNFα and IL-1β. Findings from studies on EGCG presented here may be useful in the development of more effective anti-MMP agents, which would give beneficial effects on cancer and other diseases.
Collapse
Affiliation(s)
- Hiroki Tanabe
- Faculty of Health and Welfare Science, Nayoro City University, Nayoro 096-8641, Hokkaido, Japan
- Correspondence: (H.T.); (T.O.)
| | - Takuji Suzuki
- Department of Food Science and Nutrition, Faculty of Human Life and Science, Doshisha Women’s College of Liberal Arts, Kyoto 602-0893, Japan
| | - Tomokazu Ohishi
- Institute of Microbial Chemistry (BIKAKEN), Numazu, Microbial Chemistry Research Foundation, Numazu 410-0301, Shizuoka, Japan
- Institute of Microbial Chemistry (BIKAKEN), Laboratory of Oncology, Microbial Chemistry Research Foundation, Shinagawa, Tokyo 141-0021, Japan
- Correspondence: (H.T.); (T.O.)
| | - Mamoru Isemura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Yoriyuki Nakamura
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| | - Keiko Unno
- Tea Science Center, University of Shizuoka, Suruga-ku, Shizuoka 422-8526, Japan
| |
Collapse
|
4
|
Hu F, Chen J, Zhang Y, Xu K, Sun Y, Liu Y, Cai H. MiR-219a-5p Inhibits Michigan Cancer Foundation-7 (MCF-7) Cell Migration by Regulating Zinc Finger E-Box Binding Homeobox 1 (ZEB1) Expression. J BIOMATER TISS ENG 2022. [DOI: 10.1166/jbt.2022.3135] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/23/2022]
Abstract
The potential role of miR-219a-5p in the migration of breast cancer has not been fully elucidated. In this study, bioinformatic analysis revealed that high miR-219a-5p expression in breast cancer tissue was associated with good survival of breast cancer patients. RT-qPCR analysis indicated
that miR-219a-5p expression is significantly lower in MDA-MB-231 triple-negative breast cancer (TNBC) cells. In addition, pre-miR-219a overexpression inhibited MCF-7 cell migration and inhibited ZEB1, Twist1 and vimentin expression but promoted the expression of E-cadherin. Moreover, miR-219a-5p
mimics inhibited MCF-7 cell migration, whereas MCF-7 cell migration was promoted by the miR-219a-5p inhibitor. Furthermore, miR-219a-5p was found to inhibit the translation of ZEB1 expression by targeting the 5′-ACAAUCA-3′ motif of the ZEB1 3′UTR, and the binding motif is
conserved in multiple species. ZEB1 overexpression rescued the inhibition of cell migration induced by miR-219a-5p. Finally, an inverse correlation of miR-219a-5p and ZEB1 expression was observed in four breast cancer cell lines. Thus, miR-219a-5p inhibits MCF-7 cell migration by regulating
ZEB1 expression, and ZEB1 is the target gene of miR-219a-5p.
Collapse
Affiliation(s)
- Fen Hu
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P. R. China
| | - Jiaxin Chen
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P. R. China
| | - Yunfeng Zhang
- Department of Life Sciences, Tangshan Normal University, Tangshan, Hebei 063000, P. R. China
| | - Ke Xu
- Department of Life Sciences, Tangshan Normal University, Tangshan, Hebei 063000, P. R. China
| | - Yuxi Sun
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P. R. China
| | - Yan Liu
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P. R. China
| | - Haifeng Cai
- The Second Department of Breast Surgery, Tangshan People’s Hospital, Tangshan, Hebei 063000, P. R. China
| |
Collapse
|
5
|
BMP2 as a promising anticancer approach: functions and molecular mechanisms. Invest New Drugs 2022; 40:1322-1332. [PMID: 36040572 DOI: 10.1007/s10637-022-01298-4] [Citation(s) in RCA: 15] [Impact Index Per Article: 5.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 07/09/2022] [Accepted: 08/22/2022] [Indexed: 10/14/2022]
Abstract
Bone morphogenetic protein 2 (BMP2), a pluripotent factor, is a member of the transforming growth factor-beta (TGF-β) superfamily and is implicated in embryonic development and postnatal homeostasis in tissues and organs. Experimental research in the contexts of physiology and pathology has indicated that BMP2 can induce macrophages to differentiate into osteoclasts and accelerate the osteolytic mechanism, aggravating cancer cell bone metastasis. Emerging studies have stressed the potent regulatory effect of BMP2 in cancer cell differentiation, proliferation, survival, and apoptosis. Complicated signaling networks involving multiple regulatory proteins imply the significant biological functions of BMP2 in cancer. In this review, we comprehensively summarized and discussed the current evidence related to the modulation of BMP2 in tumorigenesis and development, including evidence related to the roles and molecular mechanisms of BMP2 in regulating cancer stem cells (CSCs), epithelial-mesenchymal transition (EMT), cancer angiogenesis and the tumor microenvironment (TME). All these findings suggest that BMP2 may be an effective therapeutic target for cancer and a new marker for assessing treatment efficacy.
Collapse
|
6
|
Xu Y, Shu D, Shen M, Wu Q, Peng Y, Liu L, Tang Z, Gao S, Wang Y, Liu S. Development and Validation of a Novel PPAR Signaling Pathway-Related Predictive Model to Predict Prognosis in Breast Cancer. J Immunol Res 2022; 2022:9412119. [PMID: 35692496 PMCID: PMC9184151 DOI: 10.1155/2022/9412119] [Citation(s) in RCA: 8] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/24/2022] [Accepted: 05/06/2022] [Indexed: 12/27/2022] Open
Abstract
This study is aimed at exploring the potential mechanism of the PPAR signaling pathway in breast cancer (BRCA) and constructing a novel prognostic-related risk model. We used various bioinformatics methods and databases to complete our exploration in this research. Based on TCGA database, we use multiple extension packages based on the R language for data conversion, processing, and statistics. We use LASSO regression analysis to establish a prognostic-related risk model in BRCA. And we combined the data of multiple online websites, including GEPIA, ImmuCellAI, TIMER, GDSC, and the Human Protein Atlas database to conduct a more in-depth exploration of the risk model. Based on the mRNA data in TCGA database, we conducted a preliminary screening of genes related to the PPAR signaling pathway through univariate Cox analysis, then used LASSO regression analysis to conduct a second screening, and successfully established a risk model consisting of ten genes in BRCA. The results of ROC curve analysis show that the risk model has good prediction accuracy. We can successfully divide breast cancer patients into high- and low-risk groups with significant prognostic differences (P = 1.92e - 05) based on this risk model. Combined with the clinical data in TCGA database, there is a correlation between the risk model and the patient's N, T, gender, and fustat. The results of multivariate Cox regression show that the risk score of this risk model can be used as an independent risk factor for BRCA patients. In particular, we draw a nomogram that can predict the 5-, 7-, and 10-year survival rates of BRCA patients. Subsequently, we conducted a series of pancancer analyses of CNV, SNV, OS, methylation, and immune infiltration for this risk model gene and used GDSC data to investigate drug sensitivity. Finally, to gain insight into the predictive value and protein expression of these risk model genes in breast cancer, we used GEO and HPA databases for validation. This study provides valuable clues for future research on the PPAR signaling pathway in BRCA.
Collapse
Affiliation(s)
- Yingkun Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Dan Shu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Meiying Shen
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Qiulin Wu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Li Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Zhenrong Tang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Shun Gao
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yuan Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
7
|
Xu Y, Shu D, Shen M, Wu Q, Peng Y, Liu L, Tang Z, Gao S, Wang Y, Liu S. Development and Validation of a Novel PPAR Signaling Pathway-Related Predictive Model to Predict Prognosis in Breast Cancer. J Immunol Res 2022; 2022:9412119. [PMID: 35692496 PMCID: PMC9184151 DOI: 10.1155/2022/9412119;] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [MESH Headings] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/03/2022] [Revised: 04/24/2022] [Accepted: 05/06/2022] [Indexed: 10/11/2024] Open
Abstract
This study is aimed at exploring the potential mechanism of the PPAR signaling pathway in breast cancer (BRCA) and constructing a novel prognostic-related risk model. We used various bioinformatics methods and databases to complete our exploration in this research. Based on TCGA database, we use multiple extension packages based on the R language for data conversion, processing, and statistics. We use LASSO regression analysis to establish a prognostic-related risk model in BRCA. And we combined the data of multiple online websites, including GEPIA, ImmuCellAI, TIMER, GDSC, and the Human Protein Atlas database to conduct a more in-depth exploration of the risk model. Based on the mRNA data in TCGA database, we conducted a preliminary screening of genes related to the PPAR signaling pathway through univariate Cox analysis, then used LASSO regression analysis to conduct a second screening, and successfully established a risk model consisting of ten genes in BRCA. The results of ROC curve analysis show that the risk model has good prediction accuracy. We can successfully divide breast cancer patients into high- and low-risk groups with significant prognostic differences (P = 1.92e - 05) based on this risk model. Combined with the clinical data in TCGA database, there is a correlation between the risk model and the patient's N, T, gender, and fustat. The results of multivariate Cox regression show that the risk score of this risk model can be used as an independent risk factor for BRCA patients. In particular, we draw a nomogram that can predict the 5-, 7-, and 10-year survival rates of BRCA patients. Subsequently, we conducted a series of pancancer analyses of CNV, SNV, OS, methylation, and immune infiltration for this risk model gene and used GDSC data to investigate drug sensitivity. Finally, to gain insight into the predictive value and protein expression of these risk model genes in breast cancer, we used GEO and HPA databases for validation. This study provides valuable clues for future research on the PPAR signaling pathway in BRCA.
Collapse
Affiliation(s)
- Yingkun Xu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Dan Shu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Meiying Shen
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Qiulin Wu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yang Peng
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Li Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Zhenrong Tang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Shun Gao
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Yuan Wang
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| | - Shengchun Liu
- Department of Endocrine and Breast Surgery, The First Affiliated Hospital of Chongqing Medical University, Chongqing 400042, China
| |
Collapse
|
8
|
Sharma T, Kapoor A, Mandal CC. Duality of bone morphogenetic proteins in cancer: A comprehensive analysis. J Cell Physiol 2022; 237:3127-3163. [DOI: 10.1002/jcp.30785] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 02/14/2022] [Revised: 04/06/2022] [Accepted: 04/29/2022] [Indexed: 11/06/2022]
Affiliation(s)
- Tanu Sharma
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Anmol Kapoor
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| | - Chandi C. Mandal
- Department of Biochemistry, School of Life Sciences Central University of Rajasthan Ajmer Rajasthan India
| |
Collapse
|
9
|
Ren Y, Wang Y, Bao X, Feng M, Xing B, Lian W, Yao Y, Wang R. Diagnosis of invasive non-functional pituitary adenomas using exosomal biomarkers. Clin Chim Acta 2022; 529:25-33. [PMID: 35085587 DOI: 10.1016/j.cca.2022.01.014] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/17/2021] [Accepted: 01/20/2022] [Indexed: 11/25/2022]
Abstract
BACKGROUND AND AIMS Incomplete surgical resection of invasive non-functional pituitary adenomas (NFPAs) produces a risk of the subsequent development of complications which will require treatment with powerful drugs and adjuvant radiotherapy. MATERIALS AND METHODS The degree of invasiveness of NFPA can be established using biomarkers to help clinicians choose appropriate treatment for these patients. RESULTS This research explored transcriptomic and proteomic variations of non-invasive and invasive NFPAs, other forms of pituitary adenomas and evaluated exosomal genetic markers associated with these diseases. Increased expression of matrix metalloproteinase-1 (MMP1) and its formation in exosomes (exo-MMP1) were correlated with the characteristic invasiveness of NFPAs. Changes in the expression of MMP1 in the exosome was synchronized with transduction of NFPA cells. Enrichment of MMP1 stimulated migration, growth and angiogenesis in tumors through the protease-activated receptor-1 signaling pathway in cells. CONCLUSION The results revealed that MMP1 activity has obligatory actions in promoting tumor invasion and angiogenesis, and that the exosome-mediated regulatory pathway for MMP1 may be a novel therapeutic target.
Collapse
Affiliation(s)
- Yuan Ren
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ying Wang
- Beijing Neurosurgical Institute, Capital Medical University, Beijing 100050, China
| | - Xinjie Bao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Ming Feng
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Bing Xing
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Wei Lian
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Yong Yao
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China
| | - Renzhi Wang
- Department of Neurosurgery, Peking Union Medical College Hospital, Chinese Academy of Medical Sciences and Peking Union Medical College, Beijing 100730, China.
| |
Collapse
|
10
|
Min D, Byun J, Lee EJ, Khan AA, Liu C, Loudig O, Hu W, Zhao Y, Herlyn M, Tycko B, Cole PA, Ryu B. Epigenetic Silencing of BMP6 by the SIN3A-HDAC1/2 Repressor Complex Drives Melanoma Metastasis via FAM83G/PAWS1. Mol Cancer Res 2022; 20:217-230. [PMID: 34610961 PMCID: PMC9744461 DOI: 10.1158/1541-7786.mcr-21-0289] [Citation(s) in RCA: 1] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/21/2021] [Revised: 08/24/2021] [Accepted: 09/30/2021] [Indexed: 12/14/2022]
Abstract
Aberrant epigenetic transcriptional regulation is linked to metastasis, a primary cause of cancer-related death. Dissecting the epigenetic mechanisms controlling metastatic progression may uncover important insights to tumor biology and potential therapeutic targets. Here, we investigated the role of the SIN3A histone deacetylase 1 and 2 (SIN3A-HDAC1/2) complex in cancer metastasis. Using a mouse model of melanoma metastasis, we found that the SIN3A-HDAC1/2 transcription repressor complex silences BMP6 expression, causing increased metastatic dissemination and tumor growth via suppression of BMP6-activated SMAD5 signaling. We further discovered that FAM83G/PAWS1, a downstream effector of BMP6-SMAD5 signaling, contributes critically to metastatic progression by promoting actin-dependent cytoskeletal dynamics and cell migration. Pharmacologic inhibition of the SIN3A-HDAC1/2 complex reduced the numbers of melanoma cells in the circulation and inhibited metastatic tumor growth by inducing disseminated cell dormancy, highlighting the SIN3A-HDAC1/2 repressor complex as a potential therapeutic target for blocking cancer metastasis. IMPLICATIONS: This study identifies the novel molecular links in the metastatic progression to target cytoskeletal dynamics in melanoma and identifies the SIN3A-HDAC1/2 complex and FAM83G/PAWS1 as potential targets for melanoma adjuvant therapy.
Collapse
Affiliation(s)
- Dongkook Min
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA
| | - Jaemin Byun
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA
| | - Eun-Joon Lee
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA
| | - Abdul A Khan
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA
| | - Christina Liu
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA
| | - Oliver Loudig
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA,John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA,Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, 20057, USA
| | - Wei Hu
- Department of Chemistry and Chemistry Biology, Stevens Institute of Technology, Hoboken, NJ 07030, USA
| | - Yong Zhao
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA,John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA
| | - Meenhard Herlyn
- Molecular and Cellular Oncogenesis Program. Wistar Institute, Philadelphia, PA 19104, USA
| | - Benjamin Tycko
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA,John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA,Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, 20057, USA
| | - Phillip A Cole
- Division of Genetics, Departments of Medicine and Biological Chemistry and Molecular Pharmacology, Harvard Medical School and Brigham and Women’s Hospital, Boston, MA 02115, USA
| | - Byungwoo Ryu
- Center for Discovery and Innovation, Hackensack University Medical Center, Nutley, NJ 07110, USA,John Theurer Cancer Center, Hackensack University Medical Center, Hackensack, NJ 07601, USA,Lombardi Comprehensive Cancer Center, Georgetown University, Washington DC, 20057, USA
| |
Collapse
|
11
|
Bashir N, Ishfaq M, Mazhar K, Khan JS, Shahid R. Upregulation of CD271 transcriptome in breast cancer promotes cell survival via NFκB pathway. Mol Biol Rep 2021; 49:487-495. [PMID: 34755264 DOI: 10.1007/s11033-021-06900-1] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 08/03/2021] [Accepted: 10/29/2021] [Indexed: 10/19/2022]
Abstract
BACKGROUND Biological treatment of many cancers currently targets membrane bound receptors located on a cell surface. We are in a great to need identify novel membrane proteins associated with migration and metastasis of breast cancer cells. CD271, a single transmembrane protein belongs to tumor necrosis factor receptor family acts and play its role in proliferation of cancer cell. The purpose of this study is to investigate the role of CD271 in breast cancer. METHODS AND RESULTS In this study we analyzed the mRNA expression of CD271 in breast tumor tissue, breast cancer cell line MCF7 and isolated cancer stem cells (MCF7-CSCs) by RT-qPCR. We also measured the protein levels through western blotting in MCF-7 cell line. CD271 was upregulated in breast cancer patients among all age groups. Within the promoter region of CD271, there is a binding site for NF-κB1 which overlaps a putative quadraplex forming sequence. While CD271 also activates NF-κB pathway, down regulation of CD271 through quadraplex targeting resulted in inhibition of NF-κB and its downstream targets Nanog and Sox2. CONCLUSION In conclusion, our data shows that CD271 and NF-κB are regulated in interdependent manner. Upon CD271 inhibition, the NF-κB expression also reduces which in turn affects the cell proliferation and migration. These results suggest that CD271 is playing a crucial rule in cancer progression by regulating NF-κB and is a good candidate for the therapeutic targeting.
Collapse
Affiliation(s)
- Nabiha Bashir
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan
| | - Mehreen Ishfaq
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan
| | - Kehkashan Mazhar
- Institute of Biomedical and Genetic Engineering (IBGE), KRL Hospital Islamabad, Islamabad, Pakistan
| | - Jahangir Sarwar Khan
- Department of General Surgery, Rawalpindi Medical University, Rawalpindi, Pakistan
| | - Ramla Shahid
- Department of Biosciences, COMSATS University Islamabad (CUI), Islamabad, Pakistan.
| |
Collapse
|
12
|
Hollenberg AM, Maqsoodi N, Phan A, Huber A, Jubril A, Baldwin AL, Yokogawa N, Eliseev RA, Mesfin A. Bone morphogenic protein-2 signaling in human disc degeneration and correlation to the Pfirrmann MRI grading system. Spine J 2021; 21:1205-1216. [PMID: 33677096 PMCID: PMC8356724 DOI: 10.1016/j.spinee.2021.03.002] [Citation(s) in RCA: 13] [Impact Index Per Article: 3.3] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Grants] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 09/18/2020] [Revised: 02/27/2021] [Accepted: 03/01/2021] [Indexed: 02/03/2023]
Abstract
BACKGROUND CONTEXT Back and neck pain secondary to disc degeneration is a major public health burden. There is a need for therapeutic treatments to restore intervertebral disc (IVD) composition and function. PURPOSE To quantify ALK3, BMP-2, pSMAD1/5/8 and MMP-13 expression in IVD specimens collected from patients undergoing surgery for disc degeneration, to correlate ALK3, BMP-2, pSMAD1/5/8 and MMP-13 expression in IVD specimens to the 5-level Pfirrmann MRI grading system, and to compare ALK3, BMP-2, pSMAD1/5/8 and MMP-13 expression between cervical and lumbar degenerative disc specimens. STUDY DESIGN An immunohistochemical study assessing ALK3, BMP-2, pSMAD1/5/8, and MMP-13 expression levels in human control and degenerative IVD specimens. METHODS Human IVD specimens were collected from surgical patients who underwent discectomy and interbody fusion at our institution between 1/2015 and 8/2017. Each patient underwent MRI prior to surgery. The degree of disc degeneration was measured according to the 5-level Pfirrmann MRI grading system. Patients were categorized into either the 1) control group (Pfirrmann grades I-II) or 2) degenerative group (Pfirrmann grades III-V). Histology slides of the collected IVD specimens were prepared and immunohistochemical staining was performed to assess ALK3, BMP-2, pSMAD1/5/8, and MMP-13 expression levels in the control and degenerative specimens. Expression levels were also correlated to the Pfirrmann criteria. Lastly, the degenerative specimens were stratified according to their vertebral level and expression levels between the degenerative lumbar and cervical discs were compared. RESULTS Fifty-two patients were enrolled; however, 2 control and 2 degenerative patients were excluded due to incomplete data sets. Of the remaining 48 patients, there were 12 control and 36 degenerative specimens. Degenerative specimens had increased expression levels of BMP-2 (p=.0006) and pSMAD1/5/8 (p<.0001). Pfirrmann grade 3 (p=.0365) and grade 4 (p=.0008) discs had significantly higher BMP-2 expression as compared to grade 2 discs. Pfirrmann grade 4 discs had higher pSMAD1/5/8 expression as compared to grade 2 discs (p<.0001). There were no differences in ALK3 or MMP-13 expression between the control and degenerative discs (p>.05). Stratifying the degenerative specimens according to their vertebral level showed no significant differences in expression levels between the lumbar and cervical discs (p>.05). CONCLUSIONS BMP-2 and pSMAD1/5/8 signaling activity was significantly upregulated in the human degenerative specimens, while ALK3 and MMP-13 expression were not significantly changed. The expression levels of BMP-2 and pSMAD1/5/8 correlate positively with the degree of disc degeneration measured according to the Pfirrmann MRI grading system. CLINICAL SIGNIFICANCE BMP-SMAD signaling represents a promising therapeutic target to restore IVD composition and function in the setting of disc degeneration.
Collapse
Affiliation(s)
- Alex M Hollenberg
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Noorullah Maqsoodi
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Amy Phan
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Aric Huber
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Ayodeji Jubril
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Avionna L Baldwin
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Noriaki Yokogawa
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Roman A Eliseev
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA
| | - Addisu Mesfin
- Department of Orthopaedics, Center for Musculoskeletal Research, University of Rochester Medical Center, Rochester, NY 14642, USA.
| |
Collapse
|
13
|
Zhang T, Liu Q, Yu M, Lan Y, Zhou J. Expression Profiles Reveal Involvement of VEGF, IGF1, BIRC5, and MMP1 in Vulvar Carcinogenesis. Technol Cancer Res Treat 2021; 20:15330338211004922. [PMID: 33888009 PMCID: PMC8071978 DOI: 10.1177/15330338211004922] [Citation(s) in RCA: 0] [Impact Index Per Article: 0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/16/2022] Open
Abstract
OBJECTIVE The objective of this study was to identify key genes and shed light on the underlying molecular mechanisms of vulvar squamous cell carcinoma (VSCC). METHODS Bioinformatic software was utilized for the identification and characterization of key differentially expressed genes (DEGs) from microarrays GSE63678 and GSE38228, which contain VSCC and normal vulvar tissue data. These microarrays were obtained from Gene Expression Omnibus (GEO). Immunohistochemical assays (55 VSCC and 50 normal vulvar tissues) were utilized to validate the expression of VEGF, IGF1, BIRC5, and MMP1 screened from the identified DEGs. SPSS 18.0 software was used for statistical analyses of the relationships between IGF1, BIRC5, VEGF, MMP1 expression levels and patient clinicopathological characteristics. RESULTS A total of 141 DEGs were identified, among which 18 genes were closely correlated with the biological characteristics of VSCC. Four of the 18 genes (VEGF, IGF1, BIRC5, and MMP1) screened from the GEO database were markedly enriched in pathways in cancer (P < 0.05), and could be considered key genes in VSCC based on KEGG (Kyoto Encyclopedia of Genes and Genomes) pathway analysis in DAVID (Database for Annotation, Visualization and Integrated Discovery).The expression levels of these 4 hub genes, determined by immunohistochemical assays, were consistent with the bioinformatics results. Higher expression of IGF1 showed significant association with well-differentiated carcinomas (P = 0.017).BIRC5 expression levels showed a positive correlation with clinical stage (P = 0.039); compared with those in menopause for over 10 years, patients in menopause for less than 10 years at the time of diagnosis tended to have significantly higher expression of BIRC5 (P = 0.003). VEGF and MMP1 expression levels were not correlated with any of the tested clinicopathological characteristics. CONCLUSION VEGF, IGF1, BIRC5, and MMP1 were identified as being associated with VSCC using integrated bioinformatic methods, which may provide important insights into the pathogenesis of this disease and help to identify new biomarkers.
Collapse
Affiliation(s)
- Tao Zhang
- Department of Gynecology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Qin Liu
- Department of Pathology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Minghua Yu
- Department of Pathology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Yibing Lan
- Department of Gynecology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| | - Jianghong Zhou
- Department of Gynecology, Women's Hospital School of Medicine Zhejiang University, Hangzhou, Zhejiang, People's Republic of China
| |
Collapse
|
14
|
Yu CL, Yu YL, Yang SF, Hsu CE, Lin CL, Hsieh YH, Chiou HL. Praeruptorin A reduces metastasis of human hepatocellular carcinoma cells by targeting ERK/MMP1 signaling pathway. ENVIRONMENTAL TOXICOLOGY 2021; 36:540-549. [PMID: 33226171 DOI: 10.1002/tox.23059] [Citation(s) in RCA: 24] [Impact Index Per Article: 6.0] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Subscribe] [Scholar Register] [Received: 11/02/2020] [Accepted: 11/08/2020] [Indexed: 06/11/2023]
Abstract
Praeruptorin A (PA) is one of the active ingredients found in the dried root of Peucedanum praeruptorum Dunn, has been reported to possess anticancer effects against various types of cancer. However, the effect of PA on human hepatocellular carcinoma (HCC) remains uncleared. In this study, our results indicated that PA did not induce cytotoxicity or alter cell cycle distribution in human HCC cells (Huh-7, SK-Hep-1, and PLC/PRF/5 cells). Instead, PA inhibited the migration and invasion of human HCC cells while downregulating the expression of matrix metalloproteinase-1 (MMP1) and activating the extracellular signal-regulated kinase (ERK) signaling pathways. Furthermore, blocking the ERK signaling pathway through siERK restored the expression of MMP1 and the invasive ability of PA-treated HCC cells. In conclusion, our results demonstrate the antimetastatic activity of PA against human HCC cells, supporting its potential as a therapeutic agent of HCC treatments.
Collapse
Affiliation(s)
- Chen-Lin Yu
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yung-Luen Yu
- Graduate Institute of Biomedical Sciences, China Medical University, Taichung, Taiwan
- Center for Molecular Medicine, China Medical University Hospital, Taichung, Taiwan
- Department of Biotechnology, Asia University, Taichung, Taiwan
| | - Shun-Fa Yang
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Chung-En Hsu
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
| | - Chia-Liang Lin
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
| | - Yi-Hsien Hsieh
- Institute of Medicine, Chung Shan Medical University, Taichung, Taiwan
- Department of Medical Research, Chung Shan Medical University Hospital, Taichung, Taiwan
| | - Hui-Ling Chiou
- School of Medical Laboratory and Biotechnology, Chung Shan Medical University, Taichung, Taiwan
- Department of Clinical Laboratory, Chung Shan Medical University Hospital, Taichung, Taiwan
| |
Collapse
|
15
|
Twisted gastrulation signaling modulator 1 promotes the ability of glioma cell through activating Akt pathway. Neuroreport 2021; 32:198-205. [PMID: 33534374 DOI: 10.1097/wnr.0000000000001581] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.5] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 02/03/2023]
Abstract
Glioblastoma is one of the most common primary nervous system tumors and has a high mortality rate. It is necessary to explore a novel biological target and treatment approach. Twisted gastrulation signaling modulator 1 (TWSG1) is expressed in many tumors and closely related to tumor growth and proliferation. However, there is almost no report about the mechanism of TWSG1 in glioma. We used a glioma chip to detect the expression level of TWSG1 by Immunohistochemistry. The overexpression and silence experiments of TWSG1 were performed to assay the biological function of TWSG1 in LN229 and U251 cells. Subcutaneous xenograft mouse model presented the effect of TWSG1 expression on the malignant behavior of tumor cells. Experimental results displayed that the expression level for TWSG1 was substantially elevated in gliomas compared to that in normal brain tissue. The expression knockdown of TWSG1 caused inhibition of glioma cell proliferation. Besides, TWSG1 overexpression enhanced proliferation in glioma cells, and the capacity of proliferation was partly abolished by the PI3K inhibitor LY294002. We found that TWSG1 affected the activity of Akt signaling pathway. In conclusion, TWSG1 is overexpressed in glioma tissue and promotes tumor proliferation through Akt signaling pathway, may serve as a potential target for glioma diagnosis and therapy.
Collapse
|
16
|
García Muro AM, García Ruvalcaba A, Rizo de la Torre LDC, Sánchez López JY. Role of the BMP6 protein in breast cancer and other types of cancer. Growth Factors 2021; 39:1-13. [PMID: 34706618 DOI: 10.1080/08977194.2021.1994964] [Citation(s) in RCA: 6] [Impact Index Per Article: 1.5] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Indexed: 01/11/2023]
Abstract
The BMP6 protein (Bone Morphogenetic Protein 6) is part of the superfamily of transforming growth factor-beta (TGF-β) ligands, participates in iron homeostasis, inhibits invasion by increasing adhesions and cell-cell type interactions and induces angiogenesis directly on vascular endothelial cells. BMP6 is coded by a tumor suppressor gene whose subexpression is related to the development and cancer progression; during neoplastic processes, methylation is the main mechanism by which gene silencing occurs. This work presents a review on the role of BMP6 protein in breast cancer (BC) and other types of cancer. The studies carried out to date suggest the participation of the BMP6 protein in the epithelial-mesenchymal transition (EMT) phenotype, cell growth and proliferation; however, these processes are affected in a variable way in the different types of cancer, the methylated CpG sites in BMP6 gene promoter, as well as the interaction with other proteins could be the cause of such variation.
Collapse
Affiliation(s)
- Andrea Marlene García Muro
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, México
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | - Azaria García Ruvalcaba
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, México
- Doctorado en Genética Humana, Centro Universitario de Ciencias de la Salud, Universidad de Guadalajara, Guadalajara, México
| | | | - Josefina Yoaly Sánchez López
- División de Genética, Centro de Investigación Biomédica de Occidente, Instituto Mexicano del Seguro Social, Guadalajara, México
| |
Collapse
|
17
|
Chen H, Pan R, Li H, Zhang W, Ren C, Lu Q, Chen H, Zhang X, Nie Y. CHRDL2 promotes osteosarcoma cell proliferation and metastasis through the BMP-9/PI3K/AKT pathway. Cell Biol Int 2021; 45:623-632. [PMID: 33245175 PMCID: PMC8049056 DOI: 10.1002/cbin.11507] [Citation(s) in RCA: 15] [Impact Index Per Article: 3.8] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 05/27/2020] [Revised: 10/13/2020] [Accepted: 11/21/2020] [Indexed: 12/11/2022]
Abstract
Various studies demonstrated that bone morphogenetic proteins (BMPs) and their antagonists contribute to the development of cancers. Chordin-like 2 (CHRDL2) is a member of BMP antagonists. However, the role and its relative mechanism of CHRDL2 in osteosarcoma remains unclear. In the present study, we demonstrated that the expression of CHRDL2 was significantly upregulated in osteosarcoma tissues and cell lines compared with adjacent tissues and human normal osteoblast. Inhibition of CHRDL2 decreased the proliferation and colony formation of osteosarcoma cells in vitro, as well as the migration and invasion. CHRDL2 overexpression induced the opposite effects. CHRDL2 can bind with BMP-9, thus decreasing BMP-9 expression and the combination to its receptor protein kinase ALK1. It was predicted that BMP-9 regulates PI3K/AKT pathways using gene set enrichment analysis. Inhibition of CHRDL2 decreased the activation of PI3K/AKT pathway, while overexpression of CHRDL2 upregulated the activation. Increasing the expression of BMP-9 reversed the effects of CHRDL2 overexpression on the activation of PI3K/AKT pathway, as well as the proliferation and metastasis of osteosarcoma cells. Take together, our present study revealed that CHRDL2 upregulated in osteosarcoma tissues and cell lines, and promoted osteosarcoma cell proliferation and metastasis through the BMP-9/PI3K/AKT pathway. CHRDL2 maybe an oncogene in osteosarcoma, as well as novel biomarker for the diagnosis of osteosarcoma.
Collapse
Affiliation(s)
- Houping Chen
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Runsang Pan
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Hao Li
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Wenguang Zhang
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Chong Ren
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Qiaoying Lu
- Department of Orthopedics, Guiyang Maternal and Child Health-Care Hospital, Guiyang, Guizhou, China
| | - Hui Chen
- Central Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Xiangyan Zhang
- Department of Respiration, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| | - Yingjie Nie
- Central Laboratory, Guizhou Provincial People's Hospital, Guiyang, Guizhou, China
| |
Collapse
|
18
|
Ma L, Zhang Y, Hu F. miR‑28‑5p inhibits the migration of breast cancer by regulating WSB2. Int J Mol Med 2020; 46:1562-1570. [PMID: 32945370 PMCID: PMC7447326 DOI: 10.3892/ijmm.2020.4685] [Citation(s) in RCA: 10] [Impact Index Per Article: 2.0] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/30/2020] [Accepted: 07/07/2020] [Indexed: 12/17/2022] Open
Abstract
MicroRNAs (miRNAs or miRs) play an important role in the tumorigenesis and progression of breast cancer. However, the function of miR‑28‑5p in breast cancer migration has yet to be determined. In the present study, Human MicroRNA Expression Database (HMED) analysis revealed that the expression level of miR‑28‑5p was significantly lower in breast cancer tissue than in normal breast tissue. Kaplan-‑Meier plotter (KMPLOT) analysis revealed that the low expression level of miR‑28‑5p was associated with a poor survival in breast cancer. In addition, reverse transcription‑quantitative PCR (RT‑qPCR) revealed that the expression of miR‑28‑5p was significantly lower in breast cancer cell lines compared with that in human mammary epithelial cells (HMECs). Moreover, transfection with miR‑28‑5p mimics suppressed the migration of MCF‑7 cells, whereas an miR‑28‑5p inhibitor exerted the opposite effect. Gene chip assay identified 648 differentially expressed genes (DEGs) in cells overexpressing miR‑28‑5p. The DEGs are enriched in the 'focal adhesion' and 'pathway in cancer' pathways. The expression levels of Ras‑related protein Rap‑1b (RAP1B), WD repeat and SOCS box containing 2 (WSB2) and vascular endothelial growth factor A (VEGFA) were confirmed by RT‑qPCR. Furthermore, transfection with miR‑28‑5p mimics decreased WSB2 expression, whereas the miR‑28‑5p inhibitor increased the expression of WSB2, at both the transcriptional and translational levels. miR‑28‑5p targets the 3'UTR of WSB2, and the binding site is conserved in multiple species, with a consensus motif of 5'‑AGCUCCUU‑3'. Moreover, WSB2 overexpression promoted the migration of MCF‑7 cells which had been inhibited by miR‑28‑5p. UALCAN analysis revealed that WSB2 was significantly upregulated in primary breast tumor tissue, and a high expression level of WSB2 was associated with a poor survival in breast cancer. Furthermore, immunohistochemistry revealed that the expression of WSB2 was markedly higher in breast cancer tissue compared with that in adjacent normal breast tissue. Taken together, the findings of the present study demonstrate that miR‑28‑5p inhibits the migration of breast cancer cells by regulating WSB2 expression, and the miR‑28‑5p/WSB2 axis may be a novel therapeutic target in breast cancer.
Collapse
Affiliation(s)
- Liang Ma
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| | - Yunfeng Zhang
- Department of Life Sciences, Tangshan Normal University, Tangshan, Hebei 063000, P.R. China
| | - Fen Hu
- College of Life Sciences, North China University of Science and Technology, Tangshan, Hebei 063210, P.R. China
| |
Collapse
|
19
|
Brunner P, Hastar N, Kaehler C, Burdzinski W, Jatzlau J, Knaus P. AMOT130 drives BMP-SMAD signaling at the apical membrane in polarized cells. Mol Biol Cell 2019; 31:118-130. [PMID: 31800378 PMCID: PMC6960409 DOI: 10.1091/mbc.e19-03-0179] [Citation(s) in RCA: 11] [Impact Index Per Article: 1.8] [Reference Citation Analysis] [Abstract] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 11/12/2022] Open
Abstract
The large isoform of the transmembrane protein angiomotin (AMOT130) controls cell proliferation and migration of many cell types. AMOT130 associates to the actin cytoskeleton and regulates tight-junction maintenance and signaling often via endosomal uptake of polarity proteins at tight junctions. AMOT130 is highly polarized and present only at the apical side of polarized cells. Here we show that bone morphogenetic protein (BMP) growth factor signaling and AMOT function are interlinked in apical-basal polarized cells. BMP6 controls AMOT internalization and endosomal trafficking in epithelial cells. AMOT130 interacts with the BMP receptor BMPR2 and facilitates SMAD activation and target gene expression. We further demonstrate that this effect of AMOT on BMP-SMAD signaling is dependent on endocytosis and specific to the apical side of polarized epithelial and endothelial cells. Knockdown of AMOT reduces SMAD signaling only from the apical side of polarized cells, while basolateral BMP-SMAD signaling is unaffected. This allows for the first time interference with BMP signaling in a polarized manner and identifies AMOT130 as a novel BMP signaling regulator.
Collapse
Affiliation(s)
- Patrizia Brunner
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.,Berlin School of Integrative Oncology, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Nurcan Hastar
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Christian Kaehler
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Wiktor Burdzinski
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany
| | - Jerome Jatzlau
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| | - Petra Knaus
- Institute of Chemistry and Biochemistry, Freie Universität Berlin, 14195 Berlin, Germany.,Berlin-Brandenburg School for Regenerative Therapies, Charité-Universitätsmedizin Berlin, 13353 Berlin, Germany
| |
Collapse
|
20
|
Abstract
Breast cancer is the most prevalent type of cancer amongst women worldwide. The mortality rate for patients with early-stage breast cancer has been decreasing, however, the 5-year survival rate for patients with metastatic disease remains poor, currently at 27%. Here, we have reviewed the current understanding of the role of bone morphogenetic protein (BMP) signaling in breast cancer progression, and have highlighted the discordant results that are reported in different studies. We propose that some of these contradictory outcomes may result from signaling through either the canonical or non-canonical pathways in different cell lines and tumors, or from different tumor-stromal interactions that occur in vivo.
Collapse
Affiliation(s)
- Lap Hing Chi
- a Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute , Heidelberg , Australia
- b School of Cancer Medicine, La Trobe University , Bundoora , Australia
| | - Allan D Burrows
- a Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute , Heidelberg , Australia
- b School of Cancer Medicine, La Trobe University , Bundoora , Australia
| | - Robin L Anderson
- a Metastasis Research Laboratory, Olivia Newton-John Cancer Research Institute , Heidelberg , Australia
- b School of Cancer Medicine, La Trobe University , Bundoora , Australia
- c Department of Clinical Pathology, The University of Melbourne , Parkville , VIC , Australia
- d Sir Peter MacCallum Department of Oncology, The University of Melbourne , Parkville , Australia
| |
Collapse
|
21
|
Suton P, Bolanca A, Grgurevic L, Erjavec I, Nikles I, Muller D, Manojlovic S, Vukicevic S, Petrovecki M, Dokuzovic S, Luksic I. Prognostic significance of bone morphogenetic protein 6 (BMP6) expression, clinical and pathological factors in clinically node-negative oral squamous cell carcinoma (OSCC). J Craniomaxillofac Surg 2018; 47:80-86. [PMID: 30503606 DOI: 10.1016/j.jcms.2018.10.003] [Citation(s) in RCA: 2] [Impact Index Per Article: 0.3] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/08/2018] [Revised: 09/14/2018] [Accepted: 10/04/2018] [Indexed: 01/27/2023] Open
Abstract
Bone morphogenetic protein 6 (BMP6) has unique properties regarding structure and function in supporting bone formation during development and adult life. Despite its known role in various malignant tumors, the prognostic significance of BMP6 expression in oral squamous cell carcinoma (OSCC) remains unknown. The aim of the study was to investigate immunohistochemical expression of BMP6 in OSCC in correlation with clinical and pathological parameters, disease recurrence and survival. In addition, we investigated other parameters in order to identify prognosticators of neck metastases and final outcome. The study included 120 patients with clinically T1-3N0 OSCC who were primarily surgically treated between 2003 and 2008. There were 99 (82.5%) male and 21 (17.5%) female patients. The five-year disease-specific survival for the whole cohort was 79.7%. Tumors smaller than 2 cm in diameter showed higher incidence of strong BMP6 expression. No statistical correlation was observed between other clinico-pathological factors and BMP6 expression. Expression of BMP6 was not associated with disease recurrence and survival. BMP6 may not serve as prognosticator of final outcome or recurrence in clinically node-negative OSCC subjects. In multivariate analysis predictors of poorer survival were positive surgical margin, moderate tumor cell differentiation and pathological involvement of levels IV and/or V.
Collapse
Affiliation(s)
- Petar Suton
- Department of Radiotherapy and Medical Oncology, University Hospital for Tumors, University Hospital Centre "Sisters of Mercy", Ilica 197, 10000, Zagreb, Croatia
| | - Ante Bolanca
- Department of Oncology and Nuclear Medicine, University Hospital Centre "Sisters of Mercy", Vinogradska Cesta 29, 10000, Zagreb, Croatia
| | - Lovorka Grgurevic
- University of Zagreb School of Medicine, Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, Salata 3, 10000, Zagreb, Croatia
| | - Igor Erjavec
- University of Zagreb School of Medicine, Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, Salata 3, 10000, Zagreb, Croatia
| | - Iva Nikles
- Department of Radiotherapy and Medical Oncology, University Hospital for Tumors, University Hospital Centre "Sisters of Mercy", Ilica 197, 10000, Zagreb, Croatia
| | - Danko Muller
- University of Zagreb School of Medicine, Department of Pathology, University Hospital Dubrava, Avenue Gojko Susak 6, 10000, Zagreb, Croatia
| | - Spomenka Manojlovic
- University of Zagreb School of Medicine, Department of Pathology, University Hospital Dubrava, Avenue Gojko Susak 6, 10000, Zagreb, Croatia
| | - Slobodan Vukicevic
- University of Zagreb School of Medicine, Laboratory of Mineralized Tissues, Center for Translational and Clinical Research, Salata 3, 10000, Zagreb, Croatia
| | - Mladen Petrovecki
- University of Rijeka School of Medicine, Department of Medical Informatics, Ulica Brace Branchetta 20, 51000, Rijeka, Croatia
| | - Stjepan Dokuzovic
- Department for Traumatology and Orthopaedic Surgery, University Hospital Dubrava, Avenue Gojko Susak 6, 10000, Zagreb, Croatia
| | - Ivica Luksic
- University of Zagreb School of Medicine, Department of Maxillofacial Surgery, University Hospital Dubrava, Avenue Gojko Susak 6, 10000, Zagreb, Croatia.
| |
Collapse
|
22
|
Stieglitz D, Lamm S, Braig S, Feuerer L, Kuphal S, Dietrich P, Arndt S, Echtenacher B, Hellerbrand C, Karrer S, Bosserhoff AK. BMP6-induced modulation of the tumor micro-milieu. Oncogene 2018; 38:609-621. [PMID: 30171260 DOI: 10.1038/s41388-018-0475-x] [Citation(s) in RCA: 19] [Impact Index Per Article: 2.7] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/09/2017] [Revised: 06/22/2018] [Accepted: 07/27/2018] [Indexed: 12/22/2022]
Abstract
Melanoma is the deadliest form of skin cancer with rising incidence, creating a significant health problem. We discovered increased expression of bone morphogenetic protein 6 (BMP6) in melanoma cells and tissues, and observed that BMP6 deficiency caused significantly delayed tumor onset and decelerated tumor progression in a melanoma mouse model. Moreover, we determined that BMP6 inhibits dermal mast cell recruitment and found that mast cell-derived mediators significantly reduced melanoma growth in vitro. In line with this, mast cell deficiency accelerated tumor onset and progression in a melanoma mouse model. Analysis of human melanoma tissues revealed a strong negative correlation between melanoma proliferation and mast cell infiltration. This study elucidates a novel role of BMP6-induced modulation of the tumor microenvironment.
Collapse
Affiliation(s)
- David Stieglitz
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Susanne Lamm
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Simone Braig
- Department of Pharmacy, Pharmaceutical Biology, University of Munich, Munich, Germany
| | - Lena Feuerer
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Silke Kuphal
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Peter Dietrich
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany
| | - Stephanie Arndt
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Bernd Echtenacher
- Regensburg Center for Interventional Immunology (RCI) and University Medical Center of Regensburg, Regensburg, Germany
| | - Claus Hellerbrand
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany.,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany
| | - Sigrid Karrer
- Department of Dermatology, University Medical Center Regensburg, Regensburg, Germany
| | - Anja-Katrin Bosserhoff
- Institute of Biochemistry, Emil Fischer Center, Department of Biochemistry and Molecular Medicine, University of Erlangen-Nürnberg, Erlangen, Germany. .,Comprehensive Cancer Center (CCC) Erlangen-EMN, Erlangen, Germany.
| |
Collapse
|
23
|
Zhang L, Ye Y, Long X, Xiao P, Ren X, Yu J. BMP signaling and its paradoxical effects in tumorigenesis and dissemination. Oncotarget 2018; 7:78206-78218. [PMID: 27661009 PMCID: PMC5363655 DOI: 10.18632/oncotarget.12151] [Citation(s) in RCA: 63] [Impact Index Per Article: 9.0] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 04/28/2016] [Accepted: 09/14/2016] [Indexed: 01/04/2023] Open
Abstract
Bone morphogenetic proteins (BMPs) play important roles in embryonic and postnatal development by regulating cell differentiation, proliferation, motility, and survival, thus maintaining homeostasis during organ and tissue development. BMPs can lead to tumorigenesis and regulate cancer progression in different stages. Therefore, we summarized studies on BMP expression, the clinical significance of BMP dysfunction in various cancer types, and the molecular regulation of various BMP-related signaling pathways. We emphasized on the paradoxical effects of BMPs on various aspects of carcinogenesis, including epithelial–mesenchymal transition (EMT), cancer stem cells (CSCs), and angiogenesis. We also reviewed the molecular mechanisms by which BMPs regulate tumor generation and progression as well as potential therapeutic targets against BMPs that might be valuable in preventing tumor growth and invasion.
Collapse
Affiliation(s)
- Lijie Zhang
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Yingnan Ye
- Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| | - Xinxin Long
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Pei Xiao
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Xiubao Ren
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China
| | - Jinpu Yu
- Department of Immunology, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Key Laboratory of Cancer Immunology and Biotherapy, Tianjin, P. R. China.,Cancer Molecular Diagnostic Center, Tianjin Medical University Cancer Institute and Hospital, National Clinical Research Center of Caner, Key Laboratory of Cancer Prevention and Therapy, Clinical Research Center for Cancer, Tianjin, P. R. China
| |
Collapse
|
24
|
Zheng LQ, Wang R, Chi SM, Li CX. Matrix metalloproteinase 1: a better biomarker for squamous cell carcinoma by multiple microarray analyses. GIORN ITAL DERMAT V 2017; 154:327-337. [PMID: 29249121 DOI: 10.23736/s0392-0488.17.05770-4] [Citation(s) in RCA: 5] [Impact Index Per Article: 0.6] [Reference Citation Analysis] [Abstract] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Indexed: 12/14/2022]
Abstract
BACKGROUND The present study aimed to validate MMP1 role in the development of squamous cell carcinoma (SCC) by bioinformatics methods. METHODS Gene expression data of 10 GSE series (5 HNSCCs and 5 cSCCs) were obtained from the Gene Expression Omnibus (GEO) database and used to identify differentially expressed genes (DEGs). RESULTS Higher expression of MMP1 was found rank number one in 9/10 GSE series of SCC. MMP1 was mainly focused on Gene Ontology (GO) terms of collagen catabolic process, extracellular matrix disassembly. The analysis results of Kyoto Encyclopedia of Genes and Genomes (KEGG) pathways mainly involved Rheumatoid arthritis, Bladder cancer and Pathways in cancer. Also, MMP1 was identified as a hub protein in the PPI network by using Cytoscape software. In addition, others MMPs members of family were analyzed. CONCLUSIONS These results suggested that MMP1 may be pivotal to the transition from normal skin to premalignant lesions to SCC, thus representing a potential therapeutic target gene of diagnosis and prevention in SCC.
Collapse
Affiliation(s)
- Li-Qiang Zheng
- Department of Dermatology, Chinese PLA General Hospital, Beijing, China.,Department of Dermatology, the 251st Hospital of Chinese PLA, Zhangjiakou, China
| | - Rui Wang
- Department of Dermatology, Chinese PLA General Hospital, Beijing, China
| | - Su-Min Chi
- Department of Physiology, Fourth Military Medical University, Xi'an, Shaanxi Province, China
| | - Cheng-Xin Li
- Department of Dermatology, Chinese PLA General Hospital, Beijing, China -
| |
Collapse
|
25
|
Zabkiewicz C, Resaul J, Hargest R, Jiang WG, Ye L. Bone morphogenetic proteins, breast cancer, and bone metastases: striking the right balance. Endocr Relat Cancer 2017; 24:R349-R366. [PMID: 28733469 PMCID: PMC5574206 DOI: 10.1530/erc-17-0139] [Citation(s) in RCA: 43] [Impact Index Per Article: 5.4] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Submit a Manuscript] [Subscribe] [Scholar Register] [Received: 06/29/2017] [Accepted: 07/21/2017] [Indexed: 12/11/2022]
Abstract
Bone morphogenetic proteins (BMPs) belong to the TGF-β super family, and are essential for the regulation of foetal development, tissue differentiation and homeostasis and a multitude of cellular functions. Naturally, this has led to the exploration of aberrance in this highly regulated system as a key factor in tumourigenesis. Originally identified for their role in osteogenesis and bone turnover, attention has been turned to the potential role of BMPs in tumour metastases to, and progression within, the bone niche. This is particularly pertinent to breast cancer, which commonly metastasises to bone, and in which studies have revealed aberrations of both BMP expression and signalling, which correlate clinically with breast cancer progression. Ultimately a BMP profile could provide new prognostic disease markers. As the evidence suggests a role for BMPs in regulating breast tumour cellular function, in particular interactions with tumour stroma and the bone metastatic microenvironment, there may be novel therapeutic potential in targeting BMP signalling in breast cancer. This review provides an update on the current knowledge of BMP abnormalities and their implication in the development and progression of breast cancer, particularly in the disease-specific bone metastasis.
Collapse
Affiliation(s)
- Catherine Zabkiewicz
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Jeyna Resaul
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Rachel Hargest
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Wen Guo Jiang
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| | - Lin Ye
- Cardiff China Medical Research CollaborativeCardiff University School of Medicine, Cardiff, UK
| |
Collapse
|
26
|
Xia S, Ji R, Xu Y, Ni X, Dong Y, Zhan W. Twisted Gastrulation BMP Signaling Modulator 1 Regulates Papillary Thyroid Cancer Cell Motility and Proliferation. J Cancer 2017; 8:2816-2827. [PMID: 28928871 PMCID: PMC5604214 DOI: 10.7150/jca.18482] [Citation(s) in RCA: 7] [Impact Index Per Article: 0.9] [Reference Citation Analysis] [Abstract] [Key Words] [Track Full Text] [Download PDF] [Figures] [Journal Information] [Subscribe] [Scholar Register] [Received: 12/24/2016] [Accepted: 05/18/2017] [Indexed: 12/02/2022] Open
Abstract
Bone morphogenetic proteins (BMPs) are growth factors that have important functions in cell proliferation, migration and differentiation. To date, BMP pathway activation has been found in multiple human tumors and is associated with enhanced malignant tumor growth and metastasis. BMP activity is tightly regulated by a family of soluble extracellular secreted BMP modulators. Twisted gastrulation BMP signaling modulator 1 (TWSG1) is a direct BMP regulator that is required for the full signaling activity of BMPs. However, the functions and mechanisms of TWSG1 in papillary thyroid cancer (PTC) metastasis have not been reported. TWSG1 expression was detected in 44 PTC tissues with lymph node metastasis (LNM) and 56 PTC tissues without LNM using quantitative real-time polymerase chain reaction (qRT-PCR). Gain- and loss-of-function approaches were used to assess the biological function of TWSG1 in PTC cells. Matrigel assays demonstrated the effect of tumor cell-derived TWSG1 on endothelial cell function. Our results showed that TWSG1 expression was significantly enhanced in PTC with LNM compared to that in PTC without LNM. TWSG1 knockdown inhibited migration, invasion and proliferation of PTC cells. Additionally, TWSG1 suppression impaired the tumor cell-induced endothelial cell sprout formation. We found that TWSG1 signaling may be transduced by the BMP target transcription factor inhibitor of DNA binding 1 (Id1) and matrix metalloproteinases (MMPs) 2 and 9. In conclusion, TWSG1 was highly expressed in metastasized PTC; tumor growth, migration and invasion were dependent on TWSG1, and it may be a new diagnostic and therapeutic target for PTC.
Collapse
Affiliation(s)
- Shujun Xia
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, 200025, P. R. of China
| | - Ri Ji
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, 200025, P. R. of China
| | - Yongmin Xu
- Department of Spine Surgery, Yijishan Hospital, the First Affiliated Hospital of Wannan Medical College, Wuhu, Anhui, 241001, P. R. of China
| | - Xiaofeng Ni
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, 200025, P. R. of China
| | - Yijie Dong
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, 200025, P. R. of China
| | - Weiwei Zhan
- Ultrasound Department, Rui Jin Hospital Shanghai Jiao Tong University School of Medicine, 197 Rui Jin Er Road, Huang Pu District, Shanghai, 200025, P. R. of China
| |
Collapse
|
27
|
Zeng YH, Zhou LY, Chen QZ, Li Y, Shao Y, Ren WY, Liao YP, Wang H, Zhu JH, Huang M, He F, Wang J, Wu K, He BC. Resveratrol inactivates PI3K/Akt signaling through upregulating BMP7 in human colon cancer cells. Oncol Rep 2017; 38:456-464. [DOI: 10.3892/or.2017.5662] [Citation(s) in RCA: 38] [Impact Index Per Article: 4.8] [Reference Citation Analysis] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 11/23/2016] [Accepted: 05/11/2017] [Indexed: 11/06/2022] Open
|
28
|
Liu M, Hu Y, Zhang MF, Luo KJ, Xie XY, Wen J, Fu JH, Yang H. MMP1 promotes tumor growth and metastasis in esophageal squamous cell carcinoma. Cancer Lett 2016; 377:97-104. [PMID: 27130665 DOI: 10.1016/j.canlet.2016.04.034] [Citation(s) in RCA: 87] [Impact Index Per Article: 9.7] [Reference Citation Analysis] [Abstract] [Key Words] [MESH Headings] [Track Full Text] [Journal Information] [Subscribe] [Scholar Register] [Received: 03/23/2016] [Revised: 04/22/2016] [Accepted: 04/22/2016] [Indexed: 11/18/2022]
Abstract
Matrix metalloproteinases play an essential role in the progression of esophageal squamous cell carcinoma (ESCC). Here, we show that MMP1 expression was markedly increased in a majority of ESCC compared with nontumorous tissue. High expressions of MMP1 were closely associated with lymph node metastasis, microvessel density and advanced TNM stage. Kaplan-Meier and multivariate analyses indicated MMP1 as an independent factor for overall survival in two independent cohorts of 613 patients with ESCC. In vitro studies demonstrated that MMP1 overexpression resulted in enhanced cell viability, abilities of colony formation and cell migration. The knockdown of MMP1 in ESCC cells resulted in the opposite phenomenon. Consistently, in vivo data showed that ectopic expression of MMP1 promoted tumor growth and metastasis. Further study revealed that MMP1 facilitated ESCC through the activation of the PI3K/AKT pathway. Inhibition of the PI3K/AKT pathway by LY294002 significantly attenuated MMP1-mediated cell proliferation and migration. Taken together, our data suggest that MMP1 functions as an oncogene and serves as a prognostic biomarker and a potential therapeutic target in ESCC.
Collapse
Affiliation(s)
- Min Liu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Ultrasound, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Yi Hu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Mei-Fang Zhang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Department of Pathology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Kong-Jia Luo
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Xiu-Ying Xie
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jing Wen
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Jian-Hua Fu
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China
| | - Hong Yang
- Sun Yat-sen University Cancer Center, State Key Laboratory of Oncology in South China, Collaborative Innovation Center for Cancer Medicine, Guangzhou, China; Guangdong Esophageal Cancer Institute, Guangzhou, China; Department of Thoracic Oncology, Sun Yat-sen University Cancer Center, Guangzhou, China.
| |
Collapse
|